Attachment 7 Literature Review

Attach_07_Literature Review.pdf

Prostate, Lung, Colorectal and Ovarian Cancer Screening Trial (PLCO) (NCI)

Attachment 7 Literature Review

OMB: 0925-0407

Document [pdf]
Download: pdf | pdf
Attachment 7
Literature Review

Literature Review
From:
Prorok PC, Andriole GL, Bresalier RS, et.al. Design of the Prostate, Lung, Colorectal and
Ovarian (PLCO) Cancer Screening Trial. Controlled Clinical Trials 21:273S-309S (2000).

Design of the PLCO Trial

275S

Ovarian (PLCO) Cancer Screening Trial is a 23-year randomized trial in which
37,000 men will be screened for prostate, lung, and colorectal cancers and 37,000
women will be screened for lung, colorectal, and ovarian cancers. Prostatespecific antigen (PSA) and digital rectal examination (DRE) (for prostate), chest
X-ray (for lung), 60-cm flexible sigmoidoscopy (for colorectal), and CA125 blood
test and transvaginal ultrasound (TVU) (for ovary) are being investigated as
screening modalities. An equal number of men and women will be followed
with routine medical care as controls. There will be a follow-up period of at
least 13 years from randomization for both intervention and control participants
to determine the effects of screening on cause-specific mortality.
This paper describes the design of this trial at the completion of protocol
development (just prior to the initiation of the pilot-phase recruitment) and
protocol modifications that have occurred since. Included are the specific rationale for each cancer site, overall design features, screening and follow-up
procedures, sample-size considerations, and data analysis plans. Recruitment
into the pilot phase began November 16, 1993, with main-phase recruitment
commencing September 30, 1994.
TRIAL RATIONALE

Prostate Cancer Screening
The DRE, the most common screening test for prostate cancer screening
prior to 1990, has never been completely evaluated. Observational studies have
examined sensitivity and case survival data, but without appropriate controls
and with no adjustment for lead-time and length biases [4, 5].
In 1984, Chodak and Schoenberg [6] reported on 811 patients from 50-80
years of age who underwent rectal examination and follow-up. Thirty-eight of
43 patients with a palpable abnormality in the prostate agreed to undergo
biopsy. The positive predictive value for prostate cancer was 29%. Forty-five
percent of the cases were stage B, 6% stage C, and 18% stage D. More recent
results from the same investigators revealed a 25% positive predictive value
with 68% of the detected tumors clinically localized [7]. Others also reported
a high proportion of localized disease when prostate cancer is detected by
routine rectal examination [8-11]. In contrast, Wajsman and Chu [12] among
others have reported that even with annual rectal examination, only 20% of
cases are localized at diagnosis. Thompson and Zeidman [13] reported that
25% of men presenting with metastatic disease had a normal prostate exam.
A summary of the data on DRE for detection of prostate cancer concluded
the following: sensitivity is 55-69%, specifiCity is 89-97%, positive predictive
value is 11-26%, and negative predictive value is 85--96% [14]. Further, the
rectal examination depends on the skill and experience of the examiner and
the presence of a cancer in the posterior prostate. However, DRE is inexpensive,
relatively noninvasive, nonmorbid, and can be taught to nonprofessional health
workers. What remains to be determined is whether routine annual screening
by rectal examination reduces prostate cancer mortality. A case-control study
involving 139 men with metastatic prostate cancer and matched controls found
the relative risk of metastatic prostate cancer to be 0.9 for men with one or
more rectal examinations compared with men with none. The 95% confidence

276S

P.C. Prorok et al.
interval was 0.5-1.7, suggesting that screening by routine DRE appears to
have little effect in detecting and treating prostate cancer before it becomes
metastatic [15].
Prostatic imaging by ultrasound, computerized tomography, and magnetic
resonance imaging have also been suggested for prostate cancer screening.
Each modality has relative advantages and disadvantages. Transrectal ultrasound has received the most attention [8, 16-22]. In a summary, Waterhouse
and Resnick [23] reported that the sensitivity and specificity of ultrasound are
too low for the procedure to be a valuable screening tool. Sensitivity ranged
from 71-92% for prostate cancer and 60-85% for subclinical disease. Specificity
ranged from 41-79%, and positive predictive values in the 30% range have
been reported. The sensitivity and positive predictive value of ultrasound may
be better than those of DRE when each is used as a single test. However, the
relatively low specificity along with the invasiveness and cost of the procedure
preclude routine screening for prostate cancer by transrectal ultrasound.
Serum PSA has been examined in several observational settings, both for
initial diagnosis of disease and as a tool to detect recurrence after initial therapy
[8, 20, 24-27]. Parameter estimates for this test include sensitivity near 70%
and positive predictive values of 17-28%, although these estimates of predictive
value are strongly dependent upon the disease prevalence in the populations
studied [28]. The potential value of PSA lies in its simplicity, objectivity, reproducibility, lack of invasiveness, and lower cost relative to ultrasound. The test
has increased the detection rate of early stage cancers, many of which may be
curable by local therapy [9, 29, 30]. However, the test must be carefully evaluated because false positives in the form of benign prostatic lesions are common,
requiring biopsies and added expense, and PSA testing cannot distinguish
between latent or biologically irrelevant versus aggressive tumors.
The use of serial tests to assess the rate of change of PSA has been evaluated
as a method to improve the specificity of the test [31]. The combination of PSA
and ultrasound has been used to determine PSA density indexed to prostate
size [32-34]. In one study, volume-adjusted PSA identified a population at
higher risk of carcinoma [35], but another study of intermediate levels of PSA
found no advantage to volume-adjusted PSA levels for screening [36]. Ratios
of free to complexed PSA can amplify the differences in PSA levels for individuals with prostate cancer versus prostatic hyperplasia [37, 38]. No statistical
advantage has been established for using the ratio of free to total PSA compared
to total PSA alone in a screened population [39]; however, the free to total PSA
ratio did improve specificity in other studies [40].
In a study by Cooner et al. [41] to resolve questions surrounding the relative
merits of the three tests, all subjects had a rectal examination, PSA determination
(Hybritech assay), and a 7-mHz ultrasound examination. Most of the participants with positive results on ultrasound plus a few other individuals were
biopsied. The pertinent findings of this study and a similar study by Lee et al.
[20] are given in Table 1. Both studies demonstrate that the rate of cancer
among subjects with positive results on ultrasonography in whom the rectal and
PSA exams are normal is extremely low. Hence, ultrasound was not included as
one of the screening tests in this trial.
Careful evaluation of prostate cancer screening is mandatory because the
natural history of the disease is variable and appropriate treatment is not clearly

277S

Design of the PLCO Trial
Table I

Effect of Rectal and Prostate-Specific Antigen Examinations on
Cancer Rate in Patients with Abnormal Rectal Ultrasound
Cooner Study
Lee Study
Biopsies
Cancer
Rate
Biopsies
Cancer
Rate

Rectal
Rectal
Rectal
Rectal

PSA
PSA
PSA
PSA

+,
+,
-,
-,

+
+
-

235
166
134
177

151
23
41
12

0.64
0.14
0.31
0.07

89
23
92
44

63
6
31
2

0.71
0.26
0.34
0.05

PSA = prostate-specificantigen.

defined [28, 42, 43]. The ~cidence of prostate cancer found at autopsy steadily
increases for each decade after age 50, and most of these lesions are clinically
latent. Some progress has been made in predicting the biologic behavior of
these tumors, but despite improved understanding of the relationship among
histologic grade, tumor volume, and biologic behavior, it is difficult to determine appropriate therapy for any given tumor [44]. A meta-analysis indicated
that patients with low-grade prostate cancer can experience long-term survival
with deferred therapy [45]. Decision analyses produce indeterminate results
because of uncertainty regarding treatment efficacy and metastatic rates for
prostate cancer [46-48]. On the other hand, a review of 60,000 cases of prostate
cancer diagnosed between 1983 and 1992 showed that men with poorly or
moderately differentiated cancer had improved survival if treated rather than
followed [49].
Screening and treatment of a large population of males could entail substantial risks and morbidity, which include urinary incontinence, urethral strictures,
sexual impotence, rectal injury, and a small probability of treatment-related
mortality [44, 50]. Given these circumstances, careful evaluation of prostate
cancer screening is needed. Currently, there is insufficient evidence with which
to decide the efficacy or effectiveness of screening asymptomatic men [44, 47].
In addition to the PLCO trial, randomized trials are underway in other countries
to address these issues [51, 52].

Lung Cancer Screening
Evaluations of chest X-ray and sputum cytology, the most common screening
tests for lung cancer, were first reported nearly 30 years ago. The early studies
include the Philadelphia Pulmonary Neoplasm Research Project [53], a nonrandomized, uncontrolled study begun in 1951; the Veterans Administration study
[54], a nonrandomized, uncontrolled study performed from 1958 to 1961; the
South London Lung Cancer Study [55], a nonrandomized, uncontrolled study
done in 1955 to 1963; the North London Cancer Study [56, 57], a randomized
study with industrial firms randomized between screening and no screening
done in the early 1960s; and the Kaiser Foundation Health Plan multiphasic
screening trial [58, 59], a controlled trial with annual chest X-ray, spirometry,
and medical questionnaire as part of the multiphasic screening begun in 1964.
None of these studies demonstrated a significant impact of screening on lung
cancer mortality. The South London study, for example, showed an increase

278S

P.C. Prorok et al.
in the survival of screen-detected cases compared with other cases found in
the same geographical region, but without adjustment for self-selection bias,
lead-time bias, overdiagnosis bias, or length bias [60, 61]. These studies typically
were small, and for most, follow-up was short, so that any small to moderate
size effect or any long-term effect was not likely to be demonstrated.
More recent studies include a randomized trial in Czechoslovakia [62, 63],
a case-control study in the former German Democratic Republic [64], and a
case-control study in Japan [65]. As with some earlier studies, the randomized
groups in the Czechoslovakian study were screened with cytology and X-ray
at two frequencies, semiannual versus every 3 years, so that there was no
unscreened control group. There was no difference in mortality between the
two groups. The German case-control study evaluated chest X-rays originally
used for control of tuberculosis. The Japanese case-control study considered
X-ray histories among deceased lung cancer cases and matched controls. In
contrast to the German study, the odds ratio of dying from lung cancer for
those screened within 12 months versus those not screened was 0.72, suggesting
some benefit from the screening.
Three other randomized controlled trials have been conducted. One trial,
the Mayo Lung Project, was initiated in 1971 for males 45 years or older who
were heavy smokers [66-68]. Participants free of lung cancer on initial screening
were randomized either to a group offered screening with sputum cytology
and chest X-ray every 4 months or to a group not offered screening but advised
to seek it annually. In the studies at the Johns Hopkins University Hospital
[69-72] and at Memorial-Sloan Kettering Cancer Center [73, 74], intervention
and control groups were offered annual chest X-ray, while the intervention
group was also offered sputum cytology every 4 months. In the Mayo Clinic
study, cases found in the screened arm were diagnosed in earlier stages than
those in the control arm. However, there was no significant reduction in lung
cancer mortality between the screened group and the control group in any of
these trials.
Therefore, at this point there is no solid evidence that screening for lung
cancer can reduce lung cancer mortality. Sputum cytology has not been shown
to be effective as an adjunct to annual chest X-ray. There is evidence that
screening with chest X-ray plus sputum cytology does improve stage at diagnosis and case survival rate relative to cases diagnosed through usual care, but
despite this there was no reduction in lung cancer mortality. However, modeling using data from these trials suggests that there may have been as much as
an 18% mortality reduction in these trials [75-77].
The Mayo study is the only one of the three which is pertinent to studying
annual X-ray in the present trial because the use of screening X-rays differed
in the two arms. However, several reservations can be noted about the Mayo
study finding. First, the study was designed to detect a 50% reduction in lung
cancer mortality and was too small to demonstrate a lesser but important
reduction of 10-15%. Second, at the time the study was terminated there were
still 40 excess cases of lung cancer in the screened group. Whether these cases
represent overdiagnosis or a screening benefit that would only be seen with
longer follow-up is not known. Third, about 50% of the men in the control
group received an annual chest X-ray [68]. Thus, the level of contamination
may have been sufficient to obscure any small to moderate benefit. Finally,

279S

Design of the PLCO Trial

Table 2 Power to Detect Various Screening Effects in Previous Studies of
Chest X-Ray Screening for Lung Cancer (Based on Actual Deaths
Observed)
Mortality Reduction (%)
Study
Philadelphia
VA
South London
North London
Kaiser
Czechoslovakia
Mayo

10

20

30

40

50

0.14
0.16
0.14
0.16
0.12
0.16
0.21

0.32
0.38
0.31
0.39
0.27
0.39
0.54

0.59
0.69
0.57
0.70
0.50
0.71
0.88

0.85
0.92
0.83
0.93
0.76
0.93
0.99

0.98
0.99
0.97
0.995
0.94
0.996
0.999

when prevalence cases were detected at the first screen, they were followed
separately and were not part of the randomized comparison. Hence, any effect
of X-ray on reducing lung cancer mortality among these cases could not have
been determined. It can also be argued that therapeutic advances may render
early detection more effective today than at the time of the Mayo trial.
The concern about insufficient size of previous studies of chest X-ray screening is illustrated in Table 2. The uncertainty in interpretation of results from
completed studies has led to differences of opinion regarding the value of
the annual chest X-ray. Whether a small but important benefit exists can be
demonstrated only by a properly designed randomized trial.

Colorectal Cancer Screening
DRE, sigmoidoscopy, and fecal occult blood testing have each been suggested
for colorectal cancer screening. However, only the fecal occult blood test has
been proven to be beneficial.
Several uncontrolled studies suggesting that the fecal occult blood test leads
to early detection have been reported [78-80] as have two case-control studies
of the effect of occult blood testing on colorectal cancer mortality. In one study,
the screening histories of fatal colorectal cancer cases and matched controls
were compared, resulting in an odds ratio of 0.69 for exposure to at least one
occult blood test over a 5-year period. The wide confidence interval (0.52-0.91)
suggested a benefit from the screening but also the need for further data [81].
In the second study, cases were less likely to have ever been screened than
controls. The odds ratio was 0.7 with a 95% confidence interval of 0.5-1.0,
consistent with a screening benefit [82].
Five prospective, controlled studies of fecal occult blood testing have also
been conducted. The Strang Clinic of New York undertook a nonrandomized
study involving some 12,000 screenees and 7000 controls designed to test the
effect of combining the stool guaiac test with annual sigmoidoscopy. Individuals were allocated to the study arms by calendar periods. A reduction in
colorectal cancer mortality of borderline significance was reported [83].
A randomized trial of the stool guaiac test began in 1974 at the University
of Minnesota, where nearly 47,000 persons ages 50-80 were randomized into
three groups: a control group, an annually screened group, and a biennially

280S

P.C. Prorok et al.
screened group. The preponderance of test slides were rehydrated. Recent
results provided the first definitive evidence that annual testing for occult blood
in the stool can reduce the death rate from colorectal cancer. The 13-year
cumulative mortality from colorectal cancer was reduced by 33% (mortality
ratio 0.67 with 95% confidence interval 0.50-0.87) [84].
A controlled trial in Nottingham, United Kingdom randomized approximately 76,000 individuals to each of two arms using lists of family practitioners.
Fecal occult blood testing every 2 years using nonrehydrated slides was offered
to the screened arm for three to six rounds of screening. A 15% reduction in
colorectal cancer mortality was reported after a median follow-up time of 7.8
years [85].
Two additional randomized trials of occult blood screening were initiated
more recently. A trial in Sweden targeted individuals in the narrow age range
of 60-64 years [86]. A Danish trial randomized about 31,000 individuals ages
45-75 into two arms. Participants in the screened arm were offered nonrehydrated fecal occult blood tests every 2 years for five rounds over a 10-year
period [87, 88]. This trial demonstrated an 18% reduction in colorectal cancer
mortality [89].
In summary, testing for occult blood in the stool as a colorectal cancer
screening maneuver has been studied in several trials, and a mortality reduction
has been demonstrated. The focus of the PLCO trial is therefore flexible sigmoidoscopy.
DRE and rigid sigmoidoscopy were both part of the multiphasic screening
program carried out by the Kaiser-Permanente Foundation, and some considered the results of this study to be evidence of the effectiveness of these tests
[90]. Approximately 5000 individuals were allocated to a study group urged
to receive an annual multiphasic checkup, and a comparable number served
as controls. After 11 years, the screened group experienced a colorectal cancer
death rate of 1.0 per 1000 participants entered compared to a rate of 3.3 per
1000 in the control group [58, 59]. The observed decrease in colorectal cancer
mortality in this study could be a real effect resulting from screening. However,
this conclusion has been questioned for several reasons [91]. Some cancers
were detected in an investigation of anemia resulting from the multiphasic
examination as well as by the two tests. Further, in a reanalysis the investigators
found that rates of sigrnoidoscopy were low in both groups (control: 25%;
screened: 30%), that there was only a slight excess of exposure to sigmoidoscopy
in the study group compared to the control group, and that there was not an
appreciable difference in removal of colorectal polyps between groups. They
concluded that this study should not be used as evidence either for or against
sigmoidoscopy screening [92]. DRE made a minor contribution. In addition, a
case-control study found no statistically significant mortality reduction from
distal rectal cancer using DRE [93].
Two additional observational cohort studies of sigmoidoscopy have been
reported. One involved 21,000 participants in Minnesota who underwent an
annual physical examination that included sigmoidoscopy [94, 95]. Polyps
discovered during screening were removed, and the number of sigmoid cancers
ultimately found was only 15% of the number expected. All of the 13 cancers
found were localized, and none of the patients had died as of 1979. The second
study followed 26,000 men and women in New York [96]. In 50 cancer patients

Design of the PLCO Trial

281S

identified by screening and followed over 15 years, the 5-year survival rate
was reported to be 90%. The interpretation that screening was of benefit in
these two studies can be questioned on several grounds. Both studies are likely
to be affected by self-selection bias of participants and by exclusion of certain
individuals from the follow-up process. In the New York study, seven people
with a history of symptoms and eight with previously diagnosed lesions were
excluded, thereby lowering the observed incidence and mortality rates. In the
Minnesota study, cases found at the initial examination were excluded from
the observed incidence, and only individuals without gastrointestinal symptoms were allowed to participate. Thus, the data cannot be validly compared
with the general population [91]. In addition, the reported survival data from
both studies are affected by lead-time and length biases, but no adjustment for
these biases was attempted.
Flexible sigmoidoscopy has been shown to be more acceptable to screenees
than rigid endoscopy, and the test appears to be very sensitive and highly
specific for cancer [97, 98]. The test can discover a high proportion of polyps,
and evidence suggests that removal of adenomas decreases the risk of colorectal
cancer [99]. The need to address the impact of flexible sigmoidoscopy screening
on colorectal cancer mortality has been discussed by several investigators [97,
100, 101]. Encouraging reports of the potential impact of this test come from
two case-control studies and from the modeling work of Eddy et al. [102, 103],
which suggests a potential mortality reduction of 25--40%. Both case-control
studies were conducted in prepaid health plans and used colorectal cancer
deaths as cases, with matched controls. Exposure to sigmoidoscopy in cases
and controls was compared [104, 105]. Rigid sigmoidoscopy was used in one
study, while a majority of the screening was by flexible sigmoidoscopy in the
other study. Both studies suggested a strong effect of sigmoidoscopy in reducing
colorectal cancer mortality, with unadjusted odds ratios of 0.30 and 0.21. The
modeling conclusions and the case-control studies are subject to the assumptions and biases in the methodologies, so that conclusive results will only be
obtained from a randomized trial.

Ovarian Cancer Screening
Traditionally, the pelvic examination has been relied on to detect ovarian
cancer, but it is insensitive to early disease and small tumors [106]. Thus, most
ovarian cancers present as late-stage disease. Two new technologies may be
useful as screening tools: CA125 and TVU.
CA125 is an antigenic determinant on a high molecular weight glycoprotein
recognized by a monoclonal antibody (OC 125) using an ovarian cell line as
an immunogen. The test is performed on peripheral blood. In mostly small
(50-150 patients) preoperative studies of women with ovarian masses, serum
CA125 levels were elevated (typically above 35 U / m L ) in 68-100% of cases
averaged over all stages and in 40-50% of stage I disease. Serum CA125 may
also be elevated with pregnancy, endometriosis, menstruation, benign ovarian
tumors, and with breast, colon, pancreatic, lung, gastric, and liver cancers [107].
CA125 was reported to have high specificity in postmenopausal women in
two prospective trials. Among 1010 postmenopausal women undergoing both
pelvic examination and CA125, the only malignancy diagnosed was detected

282S

P.C. Prorok et al.
by CA125 [107]. The specificity was 94.3%. In a study in Sweden among 5550
w o m e n over 40 years of age, nine cancers were detected, six of the nine by
CA125 [108]. Specificity was 98.5% using a threshold of 35 U / m L in w o m e n
50 years of age and older. The sensitivity of CA125 was estimated in two nested
case-control studies using sera available from two serum banks [109, 110]. The
sensitivity for a level of at least 35 U / m L ranged from 20-57% for cases occurring within the first 3 years of follow-up. These two studies also reported
a specificity of 95%.
These preoperative and prospective studies together suggest early detection
potential for CA125. However, no studies have been conducted to measure
sensitivity and specificity in a large screened population, and no randomized
trials have been initiated to assess the impact of screening with CA125 on
ovarian cancer mortality.
TVU has been proposed for ovarian cancer screening [111], but experience
with this modality is limited. In a series of 1017 tumors, 0.3% of ovarian
tumors unilocular on ultrasound were malignant, while 8% of those that were
multilocular and 39% of those that were solid were malignant [106]. Higgins
et al. and Van Nagell et al. [111, 112] have been using TVU for screening w o m e n
over the age of 40 since 1987. Using 8 cm 3 as the upper limit of normal ovarian
volume, 31 abnormal ultrasonograms (in 1000 women) were obtained; 24 of
these w o m e n u n d e r w e n t laparotomy. TVU identified all three of the cancers detected.
Estimates of yield and false positivity of ultrasound are available from several
studies of w o m e n offered periodic screening. In a cohort of 801 w o m e n ages
40-70 w h o had one or more risk factors for ovarian cancer, 163 had an abnormal
abdominal ultrasound. Surgery was performed in 30 cases, and one borderline
ovarian tumor was found [113]. In another study of abdominal ultrasound,
5479 asyrnptomatic w o m e n u n d e r w e n t periodic screening. Of 326 participants
w h o had a positive test and went on to surgery, five w o m e n were diagnosed
with stage IA or IB ovarian cancer, and four were diagnosed with metastatic
ovarian cancer [114]. TVU was also used in a study of 3220 asymptomatic,
postmenopausal women. An abnormal exam led to exploratory laparotomy in
44 women. Three primary ovarian carcinomas were found, two with stage IA
cancer [115]. Finally, both transvaginal and transabdominal ultrasound were
used to screen 1601 w o m e n with a first- or second-degree relative w h o had
ovarian cancer. There were 61 positive tests, leading to six ovarian cancers, five
stage I. There were five additional cancers, three ovarian and two peritoneal,
reported 2-44 months after the last test [116].
The available evidence is not sufficient to determine if the sensitivity and
specificity of any single ovarian cancer screening test is adequate for routine
application. The modalities m a y be complementary w h e n used together. The
cost of a test such as TVU, as well as the risks and costs associated with surgical
evaluation of any positive test result, are potential impediments to general
screening. Prospective screening trials to evaluate these modalities are required.

DESIGN FEATURES
Objectives and Global Design
The PLCO trial is designed to determine, in screenees ages 55-74 at entry,
whether:

Literature Review

- Bibliography -

304S

P.C. Prorok et al.
17. Chodak GW, Wald V, Parmer E, et al. Comparison of digital examination and
transrectal ultrasonography for the diagnosis of prostatic cancer. J Urol 1986;135:
951-954.
18. Clements R, Griffiths GJ, Peeling WB, et al. How accurate is the index finger? A
comparison of digital and ultrasound examination of the prostatic nodule. Clin
Radiol 1988;39:87-89.
19. Lee F, Littrup PJ, Torp-Pedersen ST, et al. Prostate cancer: Comparison of transrectal
US and digital rectal examination for screening. Radiology 1988;168:389-394.
20. Lee F, Torp-Pedersen S, Littrup PJ, et al. Hypoechoic lesions of the prostate: Clinical
relevance of tumor size, digital rectal examination, and prostate-specific antigen.
Radiology 1989;170:29-32.
21. McClennan BL. Transrectal US of the prostate: Is the technology leading the science?
Radiology 1988;168:571-575.
22. Torp-Pedersen ST, Littrup PJ, Lee F, et al. Early prostate cancer: Diagnostic costs of
screening transrectal US and digital rectal examination. Radiology 1988;169:351-354.
23. Waterhouse RL, Resnick MI. The use of transrectal prostatic ultrasonography in
the evaluation of patients with prostatic carcinoma. J Urol 1989;141:233-239.
24. Lange PH, Ercole CJ, Lightner DJ, et al. The value of serum prostate specific antigen
determinations before and after radical prostatectomy. J Urol 1989;141:873-879.
25. Oesterling JE, Chan DW, Epstein JI, et al. Prostate specific antigen in the preoperative and postoperative evaluation of localized prostatic cancer treated with radical
prostatectomy. J Urol 1988;139:766-772.
26. Stamey TA, Yang N, Hay AR, et al. Prostate-specific antigen as a serum marker
for adenocarcinoma of the prostate. N Engl J Med 1987;317:909-916.
27. Partin AW, Oesterling JE. The clinical usefulness of prostate specific antigen: Update 1994. J Urol 1994;152:1358--1368.
28. Coley CM, Barry MJ, Fleming C, et al. Early detection of prostate cancer. Part h
Prior probability and effectiveness of tests. The American College of Physicians.
Ann Intern Med 1997;126:394--406.
29. Brawer MK, Chetner MP, Beatie J, et al. Screening for prostatic carcinoma with
prostate specific antigen. J Urol 1992;147:841--845.
30. Catalona WJ, Smith DS, Ratliff TL, et al. Measurement of prostate-specific antigen
in serum as a screening test for prostate cancer. New Engl J Med 1991;324:1156-1161.
31. Carter HB, Pearson JD, Metter EJ, et al. Longitudinal evaluation of prostate-specific
antigen levels in men with and without prostate disease. JAMA 1992;267:2215-2220.
32. Benson MC, Whang IS, Pantuck A, et al. Prostate specific antigen density: A means
of distinguishing benign prostatic hypertrophy and prostate cancer. J Urol 1992;
147:815-816.
33. Kane RA, Littrup PJ, Babaian R, et al. Prostate-specific antigen levels in 1695 men
without evidence of prostate cancer. Findings of the American Cancer Society
National Prostate Cancer Detection Project. Cancer 1992;69:1201-1207.
34. Oesterling JE. Prostate-specific antigen: Improving its ability to diagnose early
prostate cancer. JAMA 1992;267:2236-2238.
35. Lee F, Littrup PJ, Loft-Christensen L, et al. Predicted prostate specific antigen results
using transrectal ultrasound gland volume. Differentiation of benign prostatic
hyperplasia and prostate cancer. Cancer 1992;70(Suppl 1):211-220.
36. Bangma CH, Grobbee DE, Schroder FH. Volume adjustment for intermediate prostate-specific antigen values in a screening population. Eur J Cancer 1995;31A:12-14.
37. Demura T, Watarai Y, Togashi M, et al. Measurement of prostate specific antigen
and gamma-seminoprotein ratio: A new means of distinguishing benign prostatic
hyperplasia and prostate cancer. J Urol 1993;150:1740-1745.

Design of the PLCO Trial

305S

38. Lilja H. Significance of different molecular forms of serum PSA. The free, noncomplexed form of PSA versus that complexed to alpha 1-antichymotrypsin. Urol
Clin North Am 1993;20:681-686.
39. Bangma CH, Kranse R, Blijenberg BG, et al. The value of screening tests in the
detection of prostate cancer. Part I: Results of a retrospective evaluation of 1726
men. Urology 1995;46:773--778.
40. Vashi AR, Wojno KJ, Henricks W, et al. Determination of the "reflex range" and
appropriate cutpoints for percent free prostate-specific antigen in 413 men referred
for prostatic evaluation using the AxSYM system. Urology 1997;49:19-27.
41. Cooner WH, Mosley BR, Rutherford CL Jr, et al. Prostate cancer detection in
a clinical urological practice by ultrasonography, digital rectal examination and
prostate specific antigen. J Urol 1990;143:1146-1154.
42. Johansson JE, Adami HO, Andersson SO, et al. Natural history of localised prostatic
cancer. A population-based study in 223 untreated patients. Lancet 1989;1:799-803.
43. Miller GJ. Histopathology of prostate cancer: Prediction of malignant behavior and
correlation with ultrasonography. Urology 1989;33(Suppl 6):18-26.
44. Kramer BS, Brown ML, Prorok PC, et al. Prostate cancer screening: What we know
and what we need to know. Ann Intern Med 1993;119:914-923.
45. Chodak GW, Thisted RA, Gerber GS, et al. Results of conservative management
of clinically localized prostate cancer. New Engl J Med 1994;330:242-248.
46. Fleming C, Wasson JH, Albertsen PC, et al. A decision analysis of alternative
treatment strategies for clinically localized prostate cancer. Prostate Patient Outcomes Research Team. JAMA 1993;269:2650-2658.
47. Barry MJ, Fleming C, Coley CM, et al. Should Medicare provide reimbursement
for prostate-specific antigen testing for early detection of prostate cancer? Part IV:
Estimating the risks and benefits of an early detection program. Urology 1995;
46:445-461.
48. Beck JR, Kattan MW, Miles BJ. A critique of the decision analysis for clinically
localized prostate cancer. J Urol 1994;152:1894-1899.
49. Lu-Yao GL, Yao S. Population-based study of long-term survival in patients with
clinically localised prostate cancer. Lancet 1997;349:906-910.
50. Prestigiacomo AF, Stamey TA. Physiological variation of serum prostate specific
antigen in the 4.0 to 10.0 ng./ml, range in male volunteers. J Urol 1996;155:1977-1980.
51. Schroder FH, Bangrna CH. The European randomized study of screening for prostate cancer (ERSPC). Br J Urol 1997;79(Suppl 1):68-71.
52. Auvinen A, Rietbergen JB, Denis LJ, et al. Prospective evaluation plan for randomized trials of prostate cancer screening. The International Prostate Cancer Screening
Trial Evaluation Group. J Med Screen 1996;3:97-104.
53. Boucot KR, Weiss W. Is curable lung cancer detected by semiannual screening?
JAMA 1973;224:1361-1365.
54. Lilienfeld A, Archer PG, Burnett CH, et al. An evaluation of radiologic and cytologic
screening for the early detection of lung cancer: A cooperative pilot study of the
American Cancer Society and the Veterans Administration. Cancer Res 1966;26:
2083-2121.
55. Nash FA, Morgan JM, Tomkins JG. South London lung cancer study. Br Med
J 1968;2:715-721.
56. Brett GZ. The value of lung cancer detection by six-monthly chest radiographs.
Thorax 1968;23:414--420.
57. Brett GZ. Earlier diagnosis and survival in lung cancer. Br Med J 1969;4:260-262.
58. Dales LG, Friedman GD, Collen MF. Evaluating periodic multiphasic health checkups: A controlled trial. J Chronic Dis 1979;32:385-404.

306S

P.C. Prorok et al.
59. Friedman GD, CoUen MF, Fireman BH. Multiphasic health checkup evaluation: A
16-year follow-up. J Chronic Dis 1986;39:453-463.
60. Cole P, Morrison AS. Basic issues in population screening for cancer. J Natl Cancer
Inst 1980;64:1263-1272.
61. Prorok PC, Connor RJ. Screening for the early detection of cancer. Cancer Invest
1986;4:225-238.
62. Kubik A, Polak J. Lung cancer detection. Results of a randomized prospective
study in Czechoslovakia. Cancer 1986;57:2427-2437.
63. Kubik A, Parkin DM, Khlat M, et al. Lack of benefit from semi-annual screening
for cancer of the lung: Follow-up report of a randomized controlled trial on a
population of high-risk males in Czechoslovakia. Int J Cancer 1990;45:26-33.
64. Ebeling K, Nischan P. Screening for lung cancer--Results from a case-control study.
Int J Cancer 1987;40:141-144.
65. Sobue T, Suzuki T, Naruke T. A case-control study for evaluating lung-cancer
screening in Japan. Japanese Lung-Cancer Screening Group. Int J Cancer 1992;
50:230-237.
66. Fontana RS. Early detection of lung cancer: The Mayo Lung Project. In: Prorok PC,
Miller AB, eds. Screeningfor Cancer. I---General Principles on Evaluation of Screeningfor
Cancer and Screening for Lung, Bladder and Oral Cancer, vol. 78. Geneva: UICC
Technical Report Series; 1984:107-122.
67. Fontana RS. Screening for lung cancer. In: Miller AB, ed. Screeningfor Cancer. New
York: Academic Press; 1985:377-395.
68. Fontana RS. Screening for lung cancer: Recent experience in the United States. In:
Hansen HH, ed. Lung Cancer: Basic and Clinical Aspects. Boston: Marfinus Nijhoff
Publishers; 1986:91-111.
69. Levin ML, Tockman MS, Frost JK, et al. Lung cancer mortality in males screened
by chest X-ray and cytologic sputum examination: A preliminary report. Recent
Results Cancer Res 1982;82:138-146.
70. Stitik FP, Tockman MS. Radiographic screening in the early detection of lung
cancer. Radiol Clin North Am 1978;16:347-366.
71. Stitik FP, Tockman MS, Khouri NF. Chest radiology. In: Miller AB, ed. Screening
for Cancer. New York: Academic Press; 1985:163-191.
72. Tockman MS, Frost JK, Stitik FP, et al. Screening and detection of lung cancer. In:
Aisner J, ed. Lung Cancer. New York: Churchill Livingstone; 1985:25-39.
73. Melamed MR, Flehinger BJ, Zaman MB, et al. Detection of true pathologic stage
I lung cancer in a screening program and the effect on survival. Cancer 1981;
47(Suppl 5):1182-1187.
74. Melamed MR, Flehinger BJ, Zaman MB, et al. Screening for early lung cancer.
Results of the Memorial Sloan-Kettering study in New York. Chest 1984:44-53.
75. Flehinger BJ, Kimmel M. The natural history of lung cancer in a periodically
screened population. Biometrics 1987;43:127-144.
76. Flehinger BJ, Kimmel M, Melamed MR. Natural history of adenocarcinoma-large
cell carcinoma of the lung: Conclusions from screening programs in New York
and Baltimore. J Natl Cancer Inst 1988;80:337-344.
77. Shukla R, Deddens JA, Buncher CR. Survival benefits of x-ray screening for lung
cancer after bias adjustments. Computers and Mathematics with Applications 1989;
18:937-948.
78. Friihmorgen P, Demling L. Early detection of colorectal cancer with a modified
guaiac test--A screening examination of 6,000 humans. In: Winawer S, Schottenfeld
D, Sherlock P, eds. Progress in Cancer Research and Therapy, vol. 13. Colorectal Cancer:
Prevention, Epidemiology, and Screening. New York: Raven Press; 1980:311-315.

Design of the PLCO Trial

307S

79. Winawer SJ, Andrews M, Flehinger B, et al. Progress report on controlled trial of
fecal occult blood testing for the detection of colorectal neoplasia. Cancer 1980;
45:2959-2964.
80. Winawer SJ, Fath RB Jr, Schottenfeld D, et al. Screening for colorectal cancer. In:
Miller AB, ed. Screening for Cancer. New York: Academic Press; 1985:347-366.
81. Selby JV, Friedman GD, Quesenberry CP Jr, et al. Effect of fecal occult blood testing
on mortality from colorectal cancer. A case-control study. Ann Intern Med 1993;
118:1-6.
82. Lazovich D, Weiss NS, Stevens NG, et al. A case-control study to evaluate efficacy
of screening for faecal occult blood. J Med Screen 1995;2:84-89.
83. Winawer SJ, Flehinger BJ, Schottenfeld D, et al. Screening for colorectal cancer with
fecal occult blood testing and sigmoidoscopy. J Natl Cancer Inst 1993;85:1311-1318.
84. Mandel JS, Bond JH, Church TR, et al. Reducing mortality from colorectal cancer
by screening for fecal occult blood. Minnesota Colon Cancer Control Study. N Eng
J Med 1993;328:1365--1371.
85. Hardcastle JD, Chamberlain JO, Robinson MH, et al. Randomised controlled trial
of faecal-occult-blood screening for colorectal cancer. Lancet 1996;348:1472-1477.
86. Kewenter J, Bjork S, Haglind E, et al. Screening and rescreening for colorectal
cancer. A controlled trial of fecal occult blood testing in 27,700 subjects. Cancer
1988;62:645-651.
87. Kronborg O, Fenger C, Snergaard O, et al. Initial mass screening for colorectal
cancer with fecal occult blood test. Scand J Gastroenterol 1987;22:677-686.
88. Kronborg O, Fenger C, Olsen J, et al. Repeated screening for colorectal cancer with
fecal occult blood test. A prospective randomized study at Funen, Denmark. Scand
J Gastroenterol 1989;24:599-606.
89. Kronborg O, Fenger C, Olsen J, et al. Randomized study of screening for colorectal
cancer with faecal-occult-blood test. Lancet 1996;348:1467-1471.
90. Guidelines for the cancer-related checkup: Recommendations and rationale. CA
Cancer J Clin 1980;30:194-240.
91. Morrison AS. Screening in Chronic Disease. New York: Oxford University Press; 1985.
92. Selby JV, Friedman GD, CoHen MF. Sigmoidoscopy and mortality from colorectal
cancer: The Kaiser Permanente Multiphasic Evaluation Study. J Clin Epidemiol
1988;41:427-434.
93. Herrinton LJ, Selby JV, Friedman GD, et al. Case-control study of digital-rectal
screening in relation to mortality from cancer of the distal rectum. Am J Epidemiol 1995;142:961-964.
94. Gilbertsen VA. Proctosigmoidoscopy and polypectomy in reducing the incidence
of rectal cancer. Cancer 1974;34(Suppl 3):936-939.
95. Gilbertsen VA, Nelms JM. The prevention of invasive cancer of the rectum. Cancer
1978;41:1137-1139.
96. Hertz RE. Value of periodic examinations in detecting cancer of the colon and
rectum. Postgrad Med 1960;27:290-294.
97. Crespi M, Weissman GS, Gilbertsen VA, et al. The role of proctosigmoidoscopy
in screening for colorectal neoplasia. CA Cancer J Clin 1984;34:158-166.
98. Winawer SJ, Miller C, Lightdale C, et al. Patient response to sigmoidoscopy. A
randomized, controlled trial of rigid and flexible sigmoidoscopy. Cancer 1987;60:
1905-1908.
99. Winawer SJ, Zauber AG, Ho MN, et al. Prevention of colorectal cancer by colonoscopic polypectomy. The National Polyp Study Workgroup. New Eng J Med 1993;329:
1977-1981.
100. Chamberlain J, Day NE, Hakama M, et al. UICC workshop of the Project on
Evaluation of Screening Programmes for Gastrointestinal Cancer. Int J Cancer 1986;
37:329-334.

308S

P.C. Prorok et al.
101. Neugut AI, Pita S. Role of sigmoidoscopy in screening for colorectal cancer: A
critical review. Gastroenterology 1988;95:492-499.
102. Eddy DM, Nugent FW, Eddy JF, et al. Screening for colorectal cancer in a highrisk population. Results of a mathematical model. Gastroenterology 1987;92:682-692.
103. Eddy DM. Screening for colorectal cancer. Ann Intern Med 1990;113:373-384.
104. Newcomb PA, Norfleet RG, Storer BE, et al. Screening sigmoidoscopy and colorectal
cancer mortality. J Natl Cancer Inst 1992;84:1572-1575.
105. Selby JV, Friedman GD, Quesenberry CP Jr, et al. A case-control study of screening
sigmoidoscopy and mortality from colorectal cancer. N Engl J Med 1992;
326:653-657.
106. Granberg S, Wikland M, Jansson I. Macroscopic characterization of ovarian tumors
and the relation to the histological diagnosis: Criteria to be used for ultrasound
evaluation. Gynecol Oncol 1989;35:139-144.
107. Jacobs I, Bast RC Jr. The CA 125 tumour-associated antigen: A review of the
literature. Hum Reprod 1989;4:1-12.
108. Einhorn N, Sjovall K, Knapp RC, et al. Prospective evaluation of serum CA 125
levels for early detection of ovarian cancer. Obstet Gynecol 1992;80:14-18.
109. Helzlsouer KJ, Bush TL, Alberg AJ, et al. Prospective study of serum CA-125 levels
as markers of ovarian cancer. JAMA 1993;269:1123-1126.
110. Zurawski VR Jr, Orjaseter H, Andersen A, et al. Elevated serum CA 125 levels
prior to diagnosis of ovarian neoplasia: Relevance for early detection of ovarian
cancer. Int J Cancer 1988;42:677-680.
111. Higgins RV, van Nagell JR Jr, Woods CH, et al. Interobserver variation in ovarian
measurements using transvaginal sonography. Gynecol Oncol 1990;39:69-71.
112. van Nagell JR Jr, Higgins RV, Donaldson ES, et al. Transvaginal sonography as a
screening method for ovarian cancer. A report of the first 1000 cases screened.
Cancer 1990;65:573-577.
113. Andolf E, Jorgensen C, Astedt B. Ultrasound examination for detection of ovarian
carcinoma in risk groups. Obstet Gynecol 1990;75:106-109.
114. Campbell S, Bhan V, Royston P, et al. Transabdominal ultrasound screening for
early ovarian cancer. BMJ 1989;299:1363-1367.
115. DePriest PD, van Nagell JR Jr, Gallion HH, et al. Ovarian cancer screening in
asymptomatic postmenopausal women. Gynecol Oncol 1993;51:205-209.
116. Bourne TH, Campbell S, Reynolds KM, et al. Screening for early familial ovarian
cancer with transvaginal ultrasonography and colour blood flow imaging. BMJ
1993;306:1025-1029.
117. Freedman LS, Green SB. Statistical designs for investigating several interventions
in the same study: Methods for cancer prevention trials. J Natl Cancer Inst 1990;
82:910-914.
118. Etzioni RD, Connor RJ, Prorok PC, et al. Design and analysis of cancer screening
trials. Stat Methods Med Res 1995;4:3-17.
119. Shapiro S, Venet W, Strax P, et al. Periodic Screening for Breast Cancer. The Health
Insurance Plan Project and Its Sequelae, 1963-1986. Baltimore: The Johns Hopkins
University Press; 1988.
120. Tabar L, Fagerberg G, Duffy SW, et al. Update of the Swedish two-county program
of mammographic screening for breast cancer. Radiologic Clin North Am 1992;30:
187-210.
121. Simpson NK, Johnson CC, Ogden SL, et al. Recruitment strategies in the Prostate,
Lung, Colorectal and Ovarian (PLCO) Cancer Screening Trial: The first six years.
Control Clin Trials 2000;21:356S-378S.
122. Hayes RB, Reding D, Kopp W, et al. Etiologic and early marker studies in the
Prostate, Lung, Colorectal and ovarian (PLCO) Cancer Screening Trial. Control
Clin Trials 2000;21:349S-355S.

Design of the PLCO Trial

309S

123. Weissfeld JL, Fagerstrom RM, O'Brien B. Quality control of cancer screening examination procedures in the Prostate, Lung, Colorectal and Ovarian (PLCO) Cancer
Screening Trial. Control Clin Trials 2000;21:390S-399S.
124. Miller AB, Yurgalevitch S, Weissfeld JL. Death review process in the Prostate,
Lung, Colorectal and Ovarian (PLCO) Cancer Screening Trial. Control Clin Trials
2000;21:400S-406S.
125. Taylor WF, Fontana RS. Biometric design of the Mayo Lung Project for early
detection and localization of bronchogenic carcinoma. Cancer 1972;30:1344-1347.
126. Dunnett CW, Gent M. An alternative to the use of two-sided tests in clinical trials.
Stat Med 1996;15:1729-1738.
127. 1987 Annual Cancer Statistics Review, Including Cancer Trends: 1950-1985. NIH
Publication No. 88-2789. Bethesda, MD: National Cancer Institute, Division of
Cancer Prevention and Control; 1988.
128. Potosky AL, Annett DQ, Coyle L. Guide to using CAN*TROL, version 2.0. Bethesda,
MD: National Cancer Institute, Division of Cancer Prevention and Control; 1995.
129. Brown ML, Potosky AL, Thompson GB, et al. The knowledge and use of screening
tests for colorectal and prostate cancer: Data from the 1987 National Health Interview survey. Prev Med 1990;19:562-574.
130. Polednak AP. Knowledge of colorectal cancer and use of screening tests in persons
40-74 years of age. Prev Med 1990;19:213-226.
131. Hasson MA, Fagerstrom RM, Kahane DC, et al. Design and evolution of the data
management systems in the Prostate, Lung, Colorectal and Ovarian (PLCO) Cancer
Screening Trial. Control Clin Trials 2000;21:329S-348S.
132. Day NE. Estimating the sensitivity of a screening test. J Epidemiol Community Health
1985;39:364-366.
133. Day NE, Walter SD. Simplified models of screening for chronic disease: Estimation
procedures from mass screening programs. Biometrics 1984;40:1-14.
134. Walter S, Day NE. Estimation of the duration of a pre-clinical state using screening
data. Am J Epidemiol 1983;118:865-886.
135. Kafadar K, Prorok PC. A data-analytic approach for estimating lead time and
screening benefit based on survival curves in randomized cancer screening trials.
Stat Med 1994;13:569-586.
136. Kafadar K, Prorok PC. Estimating the difference in location parameters of two
survival curves, with applications to cancer screening. J Statist Planning Inference
1997;57:165-179.
137. Day NE, Williams DRR, Khaw KT. Breast cancer screening programmes: The
development of a monitoring and evaluation system. Br J Cancer 1989;59:954-958.
138. Lan KKG, DeMets DL. Group sequential procedures: Calendar .versus information
time. Stat Med 1989;8:1191-1198.
139. Lin DY, Yao Q, Ying Z. A general theory on stochastic curtailment for censored
survival data. J Am Stat Assoc 1999;94:510-521.


File Typeapplication/pdf
File TitlePII: S0197-2456(00)00098-2
File Modified2015-03-25
File Created2005-02-17

© 2024 OMB.report | Privacy Policy